You are on page 1of 13

ARTICLE

Smart Human Serum Albumin-


Indocyanine Green Nanoparticles
Generated by Programmed Assembly
for Dual-Modal Imaging-Guided
Cancer Synergistic Phototherapy
Zonghai Sheng,†,§ Dehong Hu,†,§ Mingbin Zheng,† Pengfei Zhao,† Huilong Liu,‡ Duyang Gao,† Ping Gong,†
Guanhui Gao,† Pengfei Zhang,† Yifan Ma,*,† and Lintao Cai*,†

Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, Shenzhen Institutes of Advanced Technology, Chinese Academy of
Sciences, Shenzhen 518055, PR China and ‡Department of Oncology, General Hospital of Beijing Command, PLA, Beijing 100700, PR China. §Z. S. and D. H.
contributed equally to this paper.

ABSTRACT Phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT),
is a light-activated local treatment modality that is under intensive preclinical and clinical
investigations for cancer. To enhance the treatment efficiency of phototherapy and reduce the
light-associated side effects, it is highly desirable to improve drug accumulation and precision guided
phototherapy for efficient conversion of the absorbed light energy to reactive oxygen species (ROS) and
local hyperthermia. In the present study, a programmed assembly strategy was developed for the
preparation of human serum albumin (HSA)-indocyanine green (ICG) nanoparticles (HSA-ICG NPs) by
intermolecular disulfide conjugations. This study indicated that HSA-ICG NPs had a high accumulation with tumor-to-normal tissue ratio of 36.12 ( 5.12 at
24 h and a long-term retention with more than 7 days in 4T1 tumor-bearing mice, where the tumor and its margin, normal tissue were clearly identified via
ICG-based in vivo near-infrared (NIR) fluorescence and photoacoustic dual-modal imaging and spectrum-resolved technology. Meanwhile, HSA-ICG NPs
efficiently induced ROS and local hyperthermia simultaneously for synergetic PDT/PTT treatments under a single NIR laser irradiation. After an intravenous
injection of HSA-ICG NPs followed by imaging-guided precision phototherapy (808 nm, 0.8 W/cm2 for 5 min), the tumor was completely suppressed, no
tumor recurrence and treatments-induced toxicity were observed. The results suggest that HSA-ICG NPs generated by programmed assembly as smart
theranostic nanoplatforms are highly potential for imaging-guided cancer phototherapy with PDT/PTT synergistic effects.

KEYWORDS: indocyanine green . theranostics . photothermal therapy . photodynamic therapy . synergistic effect .
photoacoustic imaging

P
hototherapy, represented by photo- accumulation of phototherapeutic agent,
dynamic therapy (PDT) and photo- precisely focusing the laser beam on the
thermal therapy (PTT), is a non- tumor areas, and adequately converting the
invasive and effective approach for cancer absorbed light energy to ROS or heat are
treatment.1,2 This type of light-triggered important.57 At present, several types of
treatment modality has remarkably im- imaging technologies, including computed
proved selectivity and fewer side effects as tomography (CT),8 positron emission tomog-
compared to conventional radiotherapies raphy (PET),9 magnetic resonance imaging * Address correspondence to
and chemotherapies.3 In phototherapy, light (MRI),1012 ultrasonic imaging13 and photo- lt.cai@siat.ac.cn,
yf.ma@siat.ac.cn.
is administered, absorbed by the photosen- acoustic imaging (PA)14 have been applied
sitizer (PS) or photothermal agent, and con- as an assistance tool for guiding cancer Received for review August 18, 2014
verted into reactive oxygen species (ROS) or phototherapy. However, real-time and pre- and accepted December 2, 2014.
local hyperthermia, leading to tumor cell cise control of the laser irradiation scope
Published online December 02, 2014
death.4 To selectively and efficiently destroy remain a major challenge as treatment 10.1021/nn5062386
the targeted cancer cells while sparing nor- required.15 Thus, during laser treatment, it
mal tissue, sufficiently enhancing the tumor is necessary to real-time outline the tumor C 2014 American Chemical Society

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12310


www.acsnano.org
ARTICLE
Figure 1. Schematic illustration of HSA-ICG NPs for in vivo dual-modal imaging, tumor margin detection, and simultaneous
PDT/PTT treatments. Uptake of HSA-ICG NPs is presumably mediated by EPR effect (passive targeting) and the gp60
transcytosis pathway (active targeting) and subsequent binding to SPARC (secreted protein acidic and rich in cysteine, SPARC)
in the tumor cells. The tumor, tumor margin and normal tissue could be detected using in vivo NIR and PA dual-modal imaging
and spectrum-resolved technology. Upon the single NIR laser irradiation, the HSA-ICG NPs can simultaneously convert the
absorbed light energy to ROS and heat for synergistic PDT/PTT treatments.

and its margin, normal tissue, which is prone to pre- laser, and obtaining improved cancer therapy efficacy
cisely focus the laser beam on the tumor areas. Fluo- in MDA-MB-435 tumor bearing mice.23 Lin et al. con-
rescence (FL) imaging in near-infrared (NIR) window firmed that photosensitizer-loaded plasmonic vesicu-
(650900 nm) holds much promise due to minimal lar assemblies of gold nanoparticles (NPs) simulta-
autofluorescence and tissue scattering.16 It not only neously excited by 671 nm laser for PDT/PTT treat-
could real-time monitor in vivo dynamic distribution of ments, which subsequently could significantly reduce
PS or photothermal agent, but also can detect the tumor growth in vivo.24 The unique treatment im-
tumor and its margin, normal tissue using contrast proved the therapeutic efficacy, reduced operation
enhancement and spectrum-resolved technology.17 In steps and time. However, the simultaneous synergistic
addition, photoacoustic (PA) imaging has a high spatial therapy strongly depended on the overlapped both
resolution of optical imaging with the high penetration optical absorption of PS and photothermal agent. The
depth of ultrasound imaging, which can overcome particular requirement and complicated synthesis pro-
some limits of NIR FL imaging.18 Therefore, in vivo cess could limit its further applications.
NIR FL and PA dual-modal imaging combined with Indocyanine green (ICG) is a NIR dye for clinic
spectrum-resolved technology would provide a real- approved by the U.S. Food and Drug Administration
time, sensitive, noninvasive assistant tool for cancer (FDA). It not only can be used for NIR FL and PA
phototherapy. imaging, but also can convert the absorbed light
In addition, to enhance the treatment efficiency and energy to ROS and local hyperthermia for PDT and
fully exert the synergistic effect of PDT and PTT, the PS PTT, respectively.25 Therefore, ICG could be considered
and photothermal agent need to be simultaneously as a kind of ideal theranostic platform for biomedi-
delivered to tumor in a site-specific manner.1922 cal applications. Recently, several research groups
Recent studies mainly focused on the PS-photothermal including ours have developed some exogenous nano-
agent composite nanostructures for simultaneous carriers, such as polyallylamine,26 calcium phos-
PDT/PTT therapy triggered by a single NIR laser.23,24 phosilicate,2729 lipidpolymer,3033 and perfluoro-
For instance, Wang et al. prepared photosensitizer- carbon,3437 to prepare ICG NPs for improving stability
conjugated gold nanostars which could be induced and tumor-specificity. However, the tumor could
both PDT and PTT effects by single wavelength NIR be partially inhibited by the single photothermal or

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12311


www.acsnano.org
ARTICLE
Figure 2. The synthesis and characterization of HSA-ICG NPs. (a) Schematic illustration of the programmed assembly strategy
for the preparation of HSA-ICG NPs. (b) TEM image of HSA-ICG NPs, which were negatively stained with 2% phosphotungstic
acid. (c) Size distribution of HSA-ICG NPs. (d) Time-dependent temperature and (e) singlet oxygen generation of HSA-ICG NPs
during single NIR laser irradiation (808 nm laser, light dose rate: 1.0 W/cm2), CICG = 0.2 mg/mL.

photodynamic effect of ICG.29,31 Herein, a programmed for tumor imaging and its margin detection, and syner-
assembly strategy was developed for fabricating ICG gistic PDT/PTT treatments.25
NPs using endogenous human serum albumin (HSA) as
delivery system, applied for in vivo NIR FL and PA dual- RESULTS AND DISCUSSION
modal imaging, tumor margin detection and simulta- Synthesis and Characterization of the HSA-ICG NPs. HSA is
neous synergistic PDT/PTT therapy (Figure 1). The tumor the most abundant plasma protein. Each HSA molec-
could be completely ablated, and on tumor recurrence ular is composed of one sulfhydryl group and 17 pair of
was observed. The smart HSA-ICG NPs have some disulfide bonds.39 In this study, the HSA was incubated
unique advantages. (1) Biosafety: as an endogenous with excessive glutathione (GSH), an endogenous re-
protein approved by FDA for intravenous administration, ducing agent, and these disulfide bonds of HSA mo-
HSA as ICG delivery system is nontoxic, nonantigenic, lecular were cleaved. The obtained free sulfhydryl
and biodegradable.38,39 (2) Tumour targeting: HSA NPs groups were assembled again by intermolecular disul-
possess both passive and active tumor-targeting abilities fide bonds and formed HSA NPs (Figure 2a). No
via enhanced permeability and retention (EPR) effect exogenous cross-linkers and toxic chemicals were
and gp60 and SPARC receptor-mediated transcytosis, involved during the preparation process. To verify
respectively.40 (3) Stimuli response: HSA NPs are pre- our hypothesis, the changes of the total number of
pared with intermolecular cross-linking by disulfide sulfhydryl group of HSA were first monitored with
bond, which exhibit excellent reductive-sensitive Ellman's method.42 As HSA was incubated with exces-
activity.41 (4) Theranostics: HSA-ICG NPs can be used sive GSH, the average number of free sulfhydryl in each

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12312


www.acsnano.org
ARTICLE
Figure 3. Cell uptake of free ICG and HSA-ICG NPs. (a) Confocal FL images of 4T1 and 293T subcellular localization of free ICG
and HSA-ICG NPs after 3 h incubation. Blue represented FL of DAPI, and red represented FL of ICG. (b) Flow cytometric analysis
of mean FL intensity (n = 10 000 cells) in 4T1 cells incubated with media (black), free ICG (red), HSA-ICG NPs with HSA blocking
(blue) and HSA-ICG NPs (purple) for 3 h.

HSA molecular increased from 0.45 to 4.6, then decreased surrounding environment and delayed its decomposi-
to 0.5 after formation of HSA-ICG NPs (Figure S1, tion thus improving the photothermal and colloid
Supporting Information). These varies of the number stability and biocompatibility of ICG (Figure S5S7).
of sulfhydryl group indicated the formation process of Upon the NIR laser irradiation, the temperature of HSA-
intermolecular disulfide bonds, which are prone to ICG NPs increased to 59.4 °C, while the PBS only
stabilize the HSA-ICG NPs and make the NPs have increased to 26.9 °C (Figure 2d). The results suggested
reductive-sensitive activity. The circular dichroism that HSA-ICG NPs could cause significant hyperther-
(CD) spectra analysis was conducted to examine the mia, leading to an irreversible damage to tumor cells
characteristic of HSA's secondary structures (Figure S2). (above 42 °C).2 The process for photothermal response
The results illustrated that the absorbance of HSA NPs of HSA-ICG NPs was accompanied simultaneously by
in the range from 190 to 255 nm was almost same as generation of ROS. As showing Figure 2e, the FL inten-
natural HSA, indicating that the secondary structuring sity of singlet oxygen sensor (20 ,70 -dichlorofluorescin
of HSA, including R helix, β sheet and random coil, did diacetate, Figure S8) generation from HSA-ICG NPs
not change during the whole process.43 increased with prolonging the laser irradiation time,
The size of HSA-ICG NPs could be turned by chang- indicating continuous generation of singlet oxygen
ing the amount of ethanol (Figure S3). Considering the by HSA-ICG NPs. The above results indicated that
size distribution and ICG loading efficiency, the opti- HSA-ICG NPs could be used as a photothermal and
mized assembly approach was used in following ex- photodynamic agent for simultaneous PDT/PTT ther-
periments. As shown in Figure 2b, the obtained HSA- apy triggered by single NIR laser. Moreover, the gen-
ICG NPs dispersed as individual NPs with a well-defined eration of singlet oxygen of HSA-ICG NPs enhanced
spherical shape and homogeneously distributed with with temperature rise (Figure S9). The result exhibited
diameter ranging from 25 to 35 nm. The average the photothermal effect mediated by HSA-ICG NPs was
hydrodynamic diameter of HSA-ICG NPs was 75 ( benefit for PDT enhancement.44 Therefore, the simul-
2.4 nm via DLS measurement (Figure 2c), which was taneous synergistic PDT/PTT effect induced by HSA-
larger than corresponding TEM results. The reason was ICG NPs with single NIR laser could adequately convert
that DLS showed an average hydrodynamic particle the NIR laser energy to ROS and local hyperthermia for
size, while TEM images detected the dehydration enhanced phototherapy.
morphology of HSA-ICG NPs. When GSH (50 mM) was In Vitro Cell Uptake and Synergistic Phototherapy. The cell
added into HSA-ICG NPs, the average hydrodynamic uptake behavior of free ICG and HSA-ICG NPs was in-
size of HSA-ICG NPs was decreased to 8.0 ( 0.6 nm, vestigated through confocal microscopy (Figure 3a).
which indicated that prepared HSA-ICG NPs had ex- After 3 h incubation, a little amount of free ICG was
cellent reductive-sensitive activity (Figure 2c). The zeta observed in 4T1 breast cancer cells. In contrast, a large
potential of HSA-ICG NPs was 39.0 ( 0.7 mV (Figure S4). number of HSA-ICG NPs entered into cancer cells and
The content of ICG in HSA NPs was 11.0% determined localized in the cytoplasm. On the contrary, when the
by UV/vis absorption spectra. The encapsulation of HSA-ICG NPs incubated with normal cells (293T human
ICG in HSA NPs prevented its interaction with the kidney cells) under the same conditions, low FL signal

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12313


www.acsnano.org
ARTICLE
Figure 4. In vitro phototherapy. (a) FL image of 4T1 cells after PDT, PTT and simultaneous PDT/PTT treatments. Viable cells
were stained green with calcein-AM, and dead/later apoptosis cells were floating and eluted, or stained red with PI, CICG =
2.5 μg/mL. (b) Quantitative detection of 4T1 cells viability following PDT, PTT and simultaneous PDT/PTT treatments for 3 h.
The date are shown as mean ( SD (n = 3). (c) Flow cytometry analysis of 4T1 cells after PDT, PTT and simultaneous PDT/PTT
treatments. Positive PI and Annexin V-Alexa Fluor 488 cells were defined as late apoptosis/necrotic cells. CICG = 2.5 μg/mL.

was observed in the cytoplasm (Figure 3a). To further irradiation alone cannot kill cells. To achieve single PDT
demonstrate the active tumor-targeting ability of HSA- and PTT treatments, we maintained a constant tem-
ICG NPs, the competition binding experiments using perature (25 °C) or treated cells with 100 mM NaN3, a
HSA were performed. A large number of HSA-ICG NPs well-known 1O2 scavenger, in order to avoid producing
entered into 4T1 cancer cells compared to the HSA-ICG singlet oxygen during laser irradiation.21 These results
NPs with blocking group (Figure 3a). The quantitative illustrated that some cells were killed and exhibited red
results of flow cytometric analysis showed that the FL in the single PDT and PTT treated groups. On the
average FL intensity was 10.3-fold greater in HSA-ICG contrary, without any post-treatments, HSA-ICG NPs
NPs group than that in HSA-ICG NPs with HSA blocking provided simultaneously synergetic PDT/PTT to induce
group (Figure 3b). The results demonstrated that the most of cells death and exhibiting intense homo-
encapsulation of ICG in HSA NPs could improve cell geneous red FL. The quantitative treatment effect of
uptake ability, and enhance cancer cell targeting. The HSA-ICG NPs was examined by MTT assay on 4T1 cells.
reasons probably were due to the presence of albumin As shown in Figure 4b, single PDT and PTT caused 57.5
receptor, SPARC (Secreted Protein, Acidic and Rich in and 84.4% of cell dead, respectively. The simultaneous
Cysteine), in 4T1 cells. HSA-binding with SPARC and PDT/PTT treatments further induced cell death up to
subsequent uptake by tumor cells increased its accu- 92.8%. The results were further validated by flow
mulation in cancer cells.39 The enhanced cell uptake of cytometry (Figure 4c). The 4T1 cells were treated with
HSA-ICG NPs was beneficial for improving the photo- PDT, PTT, and simultaneous PDT/PTT, then labeled with
therapeutic efficiency. PI and Annexin V-Alexa Fluor 488, respectively. The
Under the NIR laser irradiation, the HSA-ICG NPs results showed that simultaneous PDT/PTT treatment
in cancer cells could induce ROS and local hyperther- more significantly induced 4T1 cell late apoptosis/
mia generation, which can be detected using DHE necrosis (94.4%) as compared with single PDT
probe and an infrared thermal camera, respectively (49.3%) or PTT (71.9%). It suggested that the simulta-
(Figure S10S11). In order to visually evaluate the neous synergistic PDT/PTT effect was responsible for
in vitro therapeutic effect of HSA-ICG NPs, the cells were the improved therapeutic efficiency of HSA-ICG NPs.
stained with calcein-AM and propidium iodide (PI) to In Vivo Dual-Modal Imaging and Tumour Margin Detection.
identify live and dead/late apoptotic cells, respectively. The feasibility of HSA-ICG NPs for in vivo NIR FL and PA
As shown in Figure 4a, cells all displayed green FL in dual-modal imaging and tumor margin detection was
the control group, which suggested that pure laser investigated. Athymic nude mice with subcutaneous

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12314


www.acsnano.org
ARTICLE
Figure 5. In vivo FL imaging of nude mice bearing 4T1 tumors after i.v. injection of free ICG and HSA-ICG NPs, respectively.
In vivo FL imaging results of (a) free ICG and (b) HSA-ICG NPs at different time intervals. (c) Ex vivo FL images of major organs
and tumors after injection of free ICG and HSA-ICG NPs at 24 h. (d) Biodistribution of free ICG and HSA-ICG NPs in mice
determined by the ICG FL from diluted tissue lysates. The data are shown as mean ( SD (n = 3).

4T1 breast cancer xenografts were used as the animal detected, and expressed as percentage of dose per
modal. When the size of tumors reached ∼60 mm3, the unit mass of tissue (% ID/g). As shown in Figure 5d, at
mice were intravenously injected with free ICG and 24 h postinjection, the ICG content in tumor reaches
HSA-ICG NPs, respectively, and acquired FL images on maximum of about 28.8% dose/g tissue, and tumor-to-
CRI maestro system using a 704 nm excitation wave- normal tissue ratio is 36.12 ( 5.12, suggesting high
length and a 735 nm filter at different time points. As tumor accumulation ratio.
shown in Figure 5a, in the case of free ICG-treated mice, As a dual-modal contrast agent, the HSA-ICG NPs
the FL signals distributed extensively in liver tissue and exhibited an excellent property of PA imaging (Figure
less in tumor tissue after 0.5 h postinjection, and S13). As showing Figure 6a, before i.v. injection of free
completely disappeared after 24 h postinjection. On ICG and HSA-ICG NPs, only large blood vessels in tumor
the contrary, the FL signal of tumor tissue strength- could be visualized using hemoglobin as an endogen-
ened with the increase of time interval, and reached a ous contrast agent.4 After i.v. injection of free ICG and
peak after 24 h postinjection of HSA-ICG NPs HSA-ICG NPs, the PA signals at tumor tissue increased
(Figure 5b). After 7 days, the FL signal in tumor tissue over time (Figure 6b). The HSA-ICG NPs exhibited
still could be detected. The pharmacokinetic data stronger PA signal than that of free ICG, which agreed
showed that the blood circulation half-life of HSA-ICG with the in vivo NIR FL imaging behavior. The enhanced
NPs was significantly prolonged compared to free ICG PA signals after 24 h postinjection visually revealed the
(t1/2 HSA-ICG NPs =2.86 h vs t1/2 free ICG =0.12 h), uniform distribution of ICG within and outside the
indicating the long blood circulation time of HSA-ICG tumor microvessels. The results showed that HSA-ICG
NPs (Figure S12). The ex vivo images of major organs NPs-based PA imaging provided a high spatial resolu-
and tumors demonstrated that most of ICG accumu- tion, which is prone to understand the distribution of
lated in the liver 24 h after i.v. injection of free ICG. The ICG in tumor tissue and get the information on tumor
HSA NPs dramatically increased the accumulation of microstructure.
ICG in tumor, followed by liver and kidneys (Figure 5c). The real-time, sensitive and long-term NIR FL and
The HSA-ICG NPs biodistribution in various tissues was PA dual-modal imaging is useful in confirming tumor

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12315


www.acsnano.org
ARTICLE
Figure 6. In vivo tumor PA imaging. (a) PA imaging of the mice bearing 4T1 tumor injected with free ICG and HSA-ICG NPs at
different times. (b) The mean PA intensity of tumor tissue at 0, 1, 6, 12, and 24 h postinjection, respectively. The data are shown
as mean ( SD (n = 3).

Figure 7. In vivo tumor margin detection. (a) Photograph showing a laser beam focusing on the tumor site. In vivo FL spectra
were obtained from the tumor site, tumor margin, and normal tissue at 785 nm excitation. (b) Photograph of tumor, tumor
margin and normal tissue labeled with different numbers. (c) Averaged FL spectra of the tumor site, tumor margin and normal
tissue. (d) Tumour mapping imaging with integrated FL intensity of HSA-ICG NPs at various locations.

location, detecting tumor large blood vessels and spatially localized phototherapy. For proof-of-concept,
optimizing the content of phototherapeutic agents.14 ICG-based FL spectrum-resolved technology provided
However, scattering of light in tumor tissue severely a promising tool for highly sensitive detection of tu-
impedes imaging of tumor margin.15 Therefore, it is mor boundary.17 785 nm NIR laser was carried out as
essential to identify the tumor margin for planning an excitation wavelength, and the in vivo emission

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12316


www.acsnano.org
ARTICLE
Figure 8. In vivo cancer phototherapy in xenograft mice models with 4T1 breast cancer. Schematic illustration shows the
tumor tissue was irradiated by NIR laser beam with three different ways: (a) part irradiation, (b) accurate irradiation, (c) over
irradiation. (d) IR thermal images of 4T1 tumor-bearing mice exposed to 808 nm laser for 5 min (0.8 W/cm2). (e) H&E stained
images of tumor sections collected from different treated groups of mice 4 h post treatment. (f) Tumour growth curves of
different groups of 4T1 tumor-bearing mice. (g) Survival rates of mice bearing 4T1 tumors after different treatments. (*)p <
0.05, (**)p < 0.01. (h) Body weights were measured during the 17 day evaluation period in mice under the different conditions.
Dates indicate means and standard errors.

spectrum of HSA-ICG NPs was obtained with spectro- Imaging-Guided In Vivo Synergistic Phototherapy. On the
meter (Figure 7a). As shown in Figure 7b, the tumor and basis of the treatment results in vitro, we expected to
its margin, normal tissue sites were first labeled with apply HSA-ICG NPs for in vivo cancer phototherapy.
different color numbers. We detected the FL spectra Under the guidance of NIR FL and PA dual-modal
of ICG at normal tissue, tumor margin and tumor posi- imaging and spectrum-resolved technology, the laser
tion using spectrum-resolved technology (Figure 7c). beam could be manipulated very precisely and flexibly,
The ICG exhibited a characteristic emission spectra and accurately focused on the whole tumor tissue
with the peak located at 816 nm. The average FL (Figure 8b). The real-time temperature change of mice
intensity of tumor positions is nearly 270 times than was analyzed using an infrared thermal camera, which
that of normal tissue. The FL intensity collected from provided an effective tool for monitoring the treat-
the tumor margin (less than 1 mm from tumor) are still ment process. As shown in Figure 8d, after the 5 min of
19 times stronger than that of normal tissue, providing NIR laser irradiation, the tumors treated with PBS
excellent delineation of the tumor. Integrated FL in- and free ICG exhibited moderate increase to 37.8 and
tensity of ICG at various locations was translated into a 40.6 °C, respectively. While the temperature of tumor
mapping imaging using origin software (Figure 7d). treated with HSA-ICG NPs increased rapidly to 57.2 °C,
The results were indeed correlated with presence of which was high enough to ablate the malignant cells.
tumor and its margin, normal tissue as determined In order to confirm the PDT effect in vivo, the laser
with bright-field image. Therefore, in vivo NIR FL irradiation was adjusted using the 5 min interval after
imaging and HSA-ICG-based spectrum-resolved tech- each minute of irradiation. The tumor exhibited a mild
nology can precisely identifying tumor position, size temperature rise to 32.4 °C, which was insufficient to
and margin, which are prone to guide focusing laser irreversibly heat damage the tumor tissue. To deter-
beam on whole tumor tissue without damaging nor- mine the antitumor effect of free ICG and HSA-ICG NPs
mal tissue. mediated by heat or ROS, H&E staining of tumor

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12317


www.acsnano.org
ARTICLE
Figure 9. In vivo cancer phototherapy in orthotopic mice models with 4T1 breast cancer. (a) IR thermal images of 4T1 tumor-
bearing mice exposed to 808 nm laser for 5 min (0.8 W/cm2). (b) Body weights were measured during the 18 day evaluation
period in mice under the different conditions. Dates indicate means and standard errors. (c) Tumour growth curves of
different groups of 4T1 orthotopic mice. (d) Survival rates of orthotopic mice bearing 4T1 tumors after different treatments.
(*)p < 0.05, (**)p < 0.01.

sections was performed. As shown in Figure 8e, in PBS tissue (Figure 8b), the growth of 4T1 tumor was com-
and free ICG treated groups, there was no obvious pletely inhibited with 100% of survival rate on day 50,
tumor necrosis in histological section. On the contrary, and there was no tumor recurrence detected. The
abundant karyolysis and sporadic necrotic was ob- simultaneous synergetic PDT/PTT therapy mediated
served in HSA-ICG NPs treated group with interval laser by HSA-ICG NPs plus single NIR laser could significantly
irradiation (PDT), and apparent extensive cancer ne- improve the phototherapeutic efficiency. During the
crosis occurred with continuous NIR laser irradiation treatments, the body weight was monitored, which
(PDT/PTT). The results indicated that simultaneous indicated the treatments-induced toxicity (Figure 8h).
PDT/PTT effect in vivo could be induced by single NIR On day 17, the weight of the control groups treated
laser irradiation. HSA-ICG NPs can coordinate PDT with with PBS and HSA-ICG NPs in mice bearing 4T1 gradu-
PTT to get higher anticancer efficiency than single PDT ally increased by 6 and 11%, and those treated with
therapy in living mice.45 HSA-ICG NPs and free ICG plus continuous or interval
In order to further investigate the antitumor effi- laser increased by 7, 8 and 11%. These groups were not
ciency of HSA-ICG NPs in mice bearing 4T1, the growth significantly different from the control group, suggest-
rate of tumors was monitored every 3 days after treat- ing that the treatment was reasonably well-tolerated.
ments (Figure 8f). The represent mice photos reflecting Moreover, the H&E staining images of major organs
the tumor size change showed in Figure S14. The collected from HSA-ICG NPs treated groups with NIR
tumors treated with PBS and HSA-ICG NPs without laser showed that neither obvious damage nor inflam-
laser irradiation and free ICG plus laser irradiation grew mation was observed compared to the control group
rapidly, indicating the 4T1 tumor growth was not (Figure S15). The results indicated that the HSA-ICG NPs
affected by HSA-ICG NPs, free ICG or laser irradiation. possessed low cytotoxicity, and being suitable for
The growth of 4T1 tumors was slightly inhibited by imaging-guided simultaneous synergistic PDT/PTT
HSA-ICG NPs plus interval and laser irradiation, which therapy.
indicated that the HSA-ICG NPs for PDT triggered by On the other hand, the continuous NIR laser beam
interval laser irradiation could partly suppress tumor would be partly or over focused on the tumor tissue
growth, but tumor regrew after 3 days post-treatment. without imaging-guided technology (Figure 8a and 8c).
The survival rate of this group was 40.0% on day 20 In HSA-ICG NPs treated groups with partly laser
post-treatment (Figure 8g). When the continuous NIR beam irradiation, the growth of 4T1 tumor was partly
laser beam was accurately focused on the whole tumor inhibited, and regrow after 6 days post-treatment

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12318


www.acsnano.org
ARTICLE
(Figure 8f). The survival rate of this group was 60% on also be successfully applied for tumor phototherapy
day 20 post-treatment (Figure 8g). On the contrary, in breast tumors in orthotopic mice models.
when the NIR laser beam was over focused on the
tumor position, the growth of 4T1 tumor was com- CONCLUSION
pletely suppressed (Figure 8f). However, the body The present study reported a programmed assembly
weight of the treated mice decreased rapidly by 23%, strategy to fabricate HSA-ICG NPs successfully for
suggesting that the treatment has high side effects in vivo dual-modal imaging and tumor margin detec-
(Figure 8g). The results demonstrated that the imaging- tion following PDT/PTT synergetic phototherapy. The
guided phototherapy was prone to accurately focus advantages of the prepared method are as follows: (1)
the NIR laser beam on the tumor and improve the It could effectively maintain the biological activity of
treatment efficiency and reduce treatments-induced HSA without using exogenous cross-linkers and toxic
toxicity. chemicals; (2) The encapsulation of ICG in HSA NPs
On the basis of the encouraging results, the HSA- remarkably improved the stability of ICG; and (3) HSA
ICG NPs for synergistic phototherapy were further NPs enhanced delivery of ICG into cancer cells. The
investigated in an orthotopic breast cancer model. tumor, tumor margin and normal tissue could be
The breast tumor grew on the mammary fat pad of clearly detected using ICG-based in vivo FL and PA
mice, keeping a distance from the surface skin.46 The dual-modal imaging and spectrum-resolved technol-
mice were treated when the tumor reached 40 mm3. ogy, which was prone to avoid the effect of light
The results were shown in Figure 9. After the NIR laser scattering and precisely guide cancer phototherapy.
irradiation for 5 min, the tumors treated with HSA-ICG The HSA-ICG NPs-mediated simultaneous synergetic
NPs exhibited high increase to 53.8 °C, which was high PDT/PTT treatments significantly improved the anti-
enough to ablate the malignant cells. The growth of cancer effect, leading to superior tumor eradication
4T1 tumor in orthotopic mice models was completely without any regrowth. Therefore, this novel theraonstic
inhibited with 100% of survival rate on day 18, and nanoplatform can be a promising strategy for imaging-
there was no tumor recurrence detected (Figure S16). guided synergetic cancer phototherapy, and is ex-
The results indicated that the HSA-ICG NPs could pected to have a great potential in clinical translation.

MATERIALS AND METHODS HSA-ICG NPs Generated by Programmed Assembly. 80 mg HSA and
20 mg ICG were first dissolved in 2 mL deionized water with
Materials. HSA was obtained from Beijing Biosynthesis Bio- 50 mM GSH at 37 °C for 1 h. Then, 2 mL of the ethanol was added
technology CO., Ltd. ICG, NaN3, GSH, 3-(4,5-dimethylthiazol- into the solution to precipitate the HSA-ICG NPs. The suspension
2-yl)-2,5-diphenyltetrazolium bromide (MTT), 4,6-diamidino- was kept under stirring at room temperature for 30 min. After
2-phenylindole (DAPI), and propidium iodide (PI) were that, the suspension was dialyzed (membrane cutoff MW:
purchased from Sigma-Aldrich. Calcein-AM and Alexa Fluor 1214 KD) with deionized water at 4 °C for 24 h to remove
488 Annexin V/Propidium Iodide (PI) Cell Apoptosis Kit were ethanol, free ICG and GSH. To determine ICG loading in HSA-ICG
obtained from Invitrogen (USA). Phosphate-buffered saline NPs, the HSA-ICG NPs solution was diluted in 5 mL of DMSO/H2O
(PBS, pH 7.4), fetal bovine serum (FBS), RMPI 1640, trypsin-EDTA (9:1, v/v) and sonicated for 30 min to extract ICG completely. ICG
and penicillin-streptomycin were purchased from Gibco Life levels were determined by UV/vis absorption spectra. ICG
Technologies (AG, Switzerland). Ethanol was obtained from
loading was defined as ICG content (%, w/w) = (ICG weight in
Sinopharm Chemical Reagent Co., Ltd. (Shanghai, China). Ami-
HSA-ICG NPs/total HSA-ICG NPs weight)  100. All the measure-
con ultra-4 centrifugal filter with a molecular weight cutoff of
ments were performed in triplicate.
100 kDa was bought from Millipore (USA). All other chemicals
used in this study were of analytical reagent grade and used Characterization of HSA-ICG NPs. The freshly obtained HSA-ICG
without further purification. Superpure water (18.25 MΩ.cm, NPs suspension were transferred onto a 200 mesh copper grid
25 °C) was used to prepare all solutions. BALB/c athymic nude coated with carbon, stained with 2% (w/v) phosphotungstic
mice and BALB/c mice were maintained under aseptic condi- acid, dried at room temperature, then analyzed by TEM (FEI
tions in a small animal isolator. All food, water, bedding and Tecnai G2 F20 S-Twin, USA). UV/vis absorption spectra were
cages were autoclaved before use. obtained with a PerkinElmer Lambda 25 UV/vis spectrophot-
Determination of the Number of the Free Sulfhydryl. The number of ometer. The PL spectra were obtained by a FL spectrometer
the free sulfhydryl of HSA, reduced HSA and HSA NPs was (F900, Edinburgh Instruments Ltd.). Circular dichroism (CD)
determined using the Ellman's method.42 The preparation of spectra were recorded on an Applied Photophysis Chirascan
reduced HSA and HSA NPs was as follows. HSA was dissolved in instrument at 25 °C. The size and zeta potential of the HSA-
deionized water at concentration 40 mg/mL with 50 mM GSH at ICG NPs were measured by dynamic light scattering (DLS), using
room temperature to break up the intramolecular disulfide a Malvern Zeta Sizer (NanoZS). The samples have been
bonds and expose free sulfhydryl groups. After 60 min, 2 mL thoroughly purified with centrifugal filters from Millipore
of solution were taken out to dialyze (membrane cutoff MW: (Centrifugal filter devices, 100 K) and dispersed in superpure
12 KD) in deionized water at 4 °C for 24 h to get rid of the water prior to the measurements. Given the sensitivity of the
excessive GSH and probably its oxidized form GSSG. Then, 2 mL instrument, multiple runs (>3) were performed to avoid erro-
of the desolvating agent ethanol was added into another 2 mL neous results.
of solution to supersaturate the HSA solution and precipitate Single Oxygen Detection. 20 ,70 -dichlorofluorescin diacetate
the NPs. The suspension was kept under stirring at room was employed to evaluate the singlet oxygen generation of
temperature for 30 min. After that, the suspension was dialyzed HSA-ICG NPs. A certain of HSA-ICG NPs was mixed with 20 ,70 -
(membrane cutoff MW: 1214 KD) with deionized water at 4 °C dichlorofluorescin diacetate (1 μM) in water containing 2%
for 24 h to remove ethanol and GSH. DMSO. The mixture solutions were irradiated with an NIR laser

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12319


www.acsnano.org
ARTICLE
(808 nm, 1 W/cm2). The fluorescence was excited with a light Animals. The procedures were approved by Shenzhen Institutes
resource of 494 nm wavelength. of Advanced Technology, Chinese Academy of Sciences Animal
Cell Culture. The human embryonic kidney cell line 293T and Care and Use Committee. Six-to-seven week old male BALB/c
mouse breast cell line 4T1 were cultured in RMPI 1640 supple- athymic nude mice and BALB/c mice were maintained under
mented with 10% (v/v) fetal bovine serum, 1% (v/v) penicillin, aseptic conditions in a small animal isolator and were housed in
and 1% (v/v) streptomycin. Cells were incubated in a humidified a group of five in standard cages with free access to food and
incubator at 37 °C with 5% CO2. water and a 12 h light/dark cycle. All animals acclimated to the
In Vitro Cell Uptake. For in vitro studies, 1  105 4T1 and 293T animal facility for at least 7 days before experimentation. BALB/c
cells were seeded on a glass-bottomed culture dish, respec- mice subcutaneous tumor models were established by sub-
tively. After 24 h, HSA-ICG NPs or free ICG was incubated with cutaneous injection of 1  106 4T1 cells onto the hind legs of
different cells for 3 h at 37 °C. To remove the unbound con- mice. BALB/c mice orthotopic tumor models were developed as
jugates, the cells were washed three times for 10 min by using follow. 0.05 mL of the cell suspension was injected into the
shaking incubation in Tris buffer and fixed with 3.7% formalde- mammary fat pad inferior to the nipple using a 28-gauge
hyde solution (1 mL) and incubated at shaking incubation each needle. The breast tumor grew on the mammary fat pad of
for 20 min. The nuclear dye DAPI was used as a positive control mice, keeping a distance from the surface skin. All possible
to stain nuclei in the experiment. Images of cells were acquired parameters that may cause social stress, like group size, type
using a Leica DMI6000 inverted microscope with a Leica TCS SP5 (treated and nontreated), etc., among the experimental animals
confocal scanning system. were carefully monitored and avoided. Animals were observed
For cellular uptake experiment, the cells (1  105 cells per daily for any behavioral abnormalities and weighed weekly.
well) were seeded in 6-well plates and incubated overnight, and Blood Circulation. To detect the blood circulation half-life of
then incubated with 0.01 mg/mL HSA-ICG NPs or free ICG. After HSA-ICG NPs, the BALB/c mice were randomly divided into 10
incubation 3 h, cells were rinsed with PBS three times, tripsi- groups (n = 3 per group). Each group of mice was i.v. injected
nized, and resuspended with medium. Afterward, the cells were with 100 μL of HSA-ICG NPs. The blood samples with ∼100 μL
collected by Accuir C6 flow cytometer using CFlow Plus soft- were drawn from orbit of BALB/c mice postinjection of HSA-ICG
ware (BD, Ann Arbor, MI). NPs at different time points (0.1, 0.2, 0.3, 0.5, 1, 3, 6, 8, 12, 24 h).
DHE Staining. ROS formation was monitored by fluorescence Each blood sample was dissolved in 900 μL of lysis buffer. The
microscopy using dihydroethidium (DHE) as the indicator. concentration of HSA-ICG NPs in the blood was determined by
Briefly, 1  105 4T1 and 293T cells were seeded on a glass- the FL spectrum of each solubilized blood sample using a FL
bottomed culture dish. After 24 h, HSA-ICG NPs was incubated spectrometer. A series of dilutions of free ICG were measured to
with different cells for 3 h at 37 °C. After the cells were washed obtain a standard calibration curve. Blank blood sample without
3 times using PBS, 0.2 mL of DHE solution (5 μM, PBS) was added HSA-ICG NPs injection was measured to determine the blood
into the cells for 30 min at 37 °C. Afterward, the cells were rinsed autofluorescence level, which was subtracted from the FL
again with PBS, and subsequently illuminated using an 808 nm intensities of injected samples during the concentration calcu-
laser with energy density of 1 W/cm2 for 5 min. Then, the nuclear lation. The HSA-ICG NPs are presented as the percentage of
dye DAPI was used as a positive control to stain nuclei in the injected dose per gram of tissue (% ID/g).
experiment. Images of cells were acquired using a Leica In Vivo Tumour Imaging and Tumour Margin Detection. Athymic
DMI6000 inverted microscope with a Leica TCS SP5 confocal nude mice with subcutaneous 4T1 breast cancer xenografts
scanning system. were used as the animal modal. The imaging studies were
In Vitro Phototherapy. 4T1 cells (1 104 cells per well) were performed when tumors reached ∼60 mm3. We intravenously
seeded in 96-well plates and incubated overnight at 37 °C in (i.v.) injected HSA-ICG NPs or free ICG (0.5 mg ICG/kg) into mice
a humidified 5% CO2 atmosphere. After being rinsed with PBS (n = 3), and acquired FL images on CRI maestro system using a
(pH 7.4), the cells were incubated with HSA-ICG NPs for 3 h at 704 nm excitation wavelength and a 735 nm filter at different
37 °C under the same conditions. Afterward, the cells of experi- time points. The images were unmixed using the Maestro
mental group were rinsed again with PBS and immersed in software. Tumour-to-normal tissue ratio (T/N) was determined
200 μL of fresh culture medium, and subsequently illuminated and was expressed as mean ( SD. All mice were euthanized
using an 808 nm laser with energy density of 1 W/cm2 for 5 min. after the 24 h imaging. Tumours as well as major organs were
The PDT effect of HSA-ICG NPs on 4T1 cells were further verified collected and subjected to ex vivo imaging. The FL spectra of
by maintaining a constant temperature to avoid photothermal HSA-ICG NPs from several locations, including the center of the
effect, and the PTT effect of HSA-ICG NPs were further verified by tumor, tumor margin and normal tissue, were examined by
treating 4T1 cells with 100 mM NaN3 to avoid producing singlet using an NIR FL spectrometer (Inspector Series, DeltaNu). After
oxygen during laser irradiation. The laser spot was adjusted to the spectra were acquired, the integrated signal intensity was
fully cover the area of each well. After illumination, cells were calculated. In vivo PA imaging was obtained with preclinical
incubated for 12 h in a 5% CO2, 95% air humidified incubator at photoacoustic computerized tomography scanner (Endra
37 °C. Dark control group was kept under identical conditions as Nexus 128, Ann Arbor, MI).
the experimental group except for illumination. The standard In Vivo Thermal Imaging. When the tumor size reached ∼60 mm3,
MTT assay was carried out to evaluate the cell viability. HSA-ICG NPs (2 mg ICG/kg) or free ICG (2 mg ICG/kg) was
The synergistic PDT/PTT effects of HSA-ICG NPs on 4T1 cells intravenously injected into the tumor-bearing mice. Thermal
were further verified using Calcein AM and propidium iodide imaging was recorded by an infrared thermal imaging camera
(PI) costaining. 4T1 cells (5  104 cells per well) were seeded in (Ti27, Fluke, USA) when the tumors were exposed to 808 nm laser
6-well plates and incubated overnight at 37 °C in a humidified of power density at 0.8 W/cm2.
5% CO2 atmosphere. After being rinsed with PBS (pH 7.4), the In Vivo Synergistic Phototherapy. For synergistic phototherapy
cells were incubated with HSA-ICG NPs or free ICG for 3 h at studies, 35 mice bearing 4T1 tumors were randomly divided into
37 °C under the same conditions. Afterward, the cells of seven groups. The treatment scheme is as follows: (1) PBS, no irra-
experimental group were rinsed again with PBS and immersed diation; (2) HSA-ICG NPs, no irradiation; (3) free ICG (2 mg ICG/kg)
in 200 μL of fresh culture medium, and subsequently illumi- plus accurate irradiation (b); (4) HSA-ICG NPs (2 mg ICG/kg) plus
nated using an 808 nm laser with energy density of 1 W/cm2 for interval laser accurate irradiation (5 min interval after each
5 min. After another 4 h incubation cells were stained with minute of irradiation) (b); (5) HSA-ICG NPs (2 mg ICG/kg) plus
calcein-AM for visualization of live cells and with PI for visualiza- part irradiation (a); (6) HSA-ICG NPs (2 mg ICG/kg) plus over
tion of dead/late apoptotic cells, according to the manufac- irradiation (c); (7) HSA-ICG NPs (2 mg ICG/kg) plus accurate
turer's suggested protocol (Invitrogen). Afterward, the cells of irradiation (b). The photoirradiation was applied 24 h after the
experimental group were rinsed again with PBS and were injection of HSA-ICG NPs or free ICG (808 nm, 0.8 W/cm2 for
examined by eliminating the interference of background FL of 5 min). The spot of laser beam was adjusted according to the
ICG with biological inverted microscope (Olympus IX71, JPN). different experiment conditions. The tumor sizes and body
Animal Model. Animals received care in accordance with the weights were inspected every 3 days. The tumor weight was
Guidance Suggestions for the Care and Use of Laboratory estimated using the formula, tumor volume = length  (width)2/2,

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12320


www.acsnano.org
ARTICLE
assuming a tumor density of 1 mg/mL. The volume of tumors 9. Zhou, M.; Zhang, R.; Huang, M.; Lu, W.; Song, S.; Melancon,
was evaluated by normalizing the measured values. After M. P.; Tian, M.; Liang, D.; Li, C. A Chelator-Free Multifunc-
therapy, major organs as well as tumors were collected and tional [64Cu]CuS Nanoparticle Platform for Simultaneous
sectioned to 8 μm slices for H&E staining. Micro-PET/CT Imaging and Photothermal Ablation Ther-
H&E Staining. H&E staining was performed according to a apy. J. Am. Chem. Soc. 2010, 132, 15351–15358.
protocol provided by the vendor (BBC Biochemical). Briefly, 10. Hirsch, L. R.; Stafford, R. J.; Bankson, J. A.; Sershen, S. R.;
8 μm cryogenic slides were prepared and fixed with 10% formalin Rivera, B.; Price, R. E.; Hazle, J. D.; Halas, N. J.; West, J. L.
for about 30 min at room temperature. After washing with Nanoshell-Mediated Near-Infrared Thermal Therapy of
running water for 5 min, the slides were treated with gradient Tumours under Magnetic Resonance Guidance. Proc. Natl.
concentrations of alcohol (100, 95, and 70%), each for 20 s. The Acad. Sci. U. S. A. 2003, 100, 13549–13554.
hematoxylin staining was performed for about 3 min and 11. Liang, X.; Li, X.; Jing, L.; Yue, X.; Dai, Z. Theranostic
washed with water for 1 min. The eosin staining was performed Porphyrin Dyad Nanoparticles for Magnetic Resonance
for about 1 min. The slides were washed, treated with xylene, Imaging Guided Photodynamic Therapy. Biomaterials
and mounted with Canada balsam. The images were acquired 2014, 35, 6379–6388.
on a Nikon Eclipse 90i microscope. 12. Ma, Y.; Tong, S.; Bao, G.; Gao, C.; Dai, Z. Indocyanine Green
Loaded SPIO Nanoparticles with Phospholipid-PEG Coat-
Conflict of Interest: The authors declare no competing ing for Dual-Modal Imaging and Photothermal Therapy.
financial interest. Biomaterials 2013, 34, 7706–7714.
13. Ke, H.; Wang, J.; Dai, Z.; Jin, Y.; Qu, E.; Xing, Z.; Guo, C.; Yue,
Acknowledgment. We are grateful to Dr. Junhui Zhai (United X.; Liu, J. Gold-Nanoshelled Microcapsules: a Theranostic
Well Technology Limited (Shanghai)) and Professor Gang Liu Agent for Ultrasound Contrast Imaging and Photothermal
(Center for Molecular Imaging and Translational Medicine, Therapy. Angew. Chem., Int. Ed. Engl. 2011, 50, 3017–3021.
Xiamen University) for PA imaging studies. The authors grate- 14. Sheng, Z.; Song, L.; Zheng, J.; Hu, D.; He, M.; Zheng, M.; Gao,
fully acknowledge support for this research from the National G.; Gong, P.; Zhang, P.; Ma, Y.; et al. Protein-Assisted
Natural Science Foundation of China (Grant No. 81301272, Fabrication of Nano-Reduced Graphene Oxide for Com-
81401521, 81071249, 81171446 and 81371679), Science and bined In Vivo Photoacoustic Imaging and Photothermal
Technology Foundation of Guangdong Province of China (Grant Therapy. Biomaterials 2013, 34, 5236–5243.
No. 2009A030301010) and Shenzhen (JCYJ20130401170306862 15. Liang, C.; Diao, S.; Wang, C.; Gong, H.; Liu, T.; Hong, G.; Shi,
and CXZZ20130506140505859), Key Project of Science and Tech- X.; Dai, H.; Liu, Z. Tumour Metastasis Inhibition by Imaging-
nology of Guangdong (2012B090400043 and 2012B090600036), Guided Photothermal Therapy with Single-Walled Carbon
Nanshan Innovative Technology Program (Grant No. KC2013- Nanotubes. Adv. Mater. 2014, 26, 5646–5652.
JSJS0012A), Guangdong Innovation Team of Low-cost Health- 16. Frangioni, J. V. In Vivo Near-Infrared Fluorescence Imaging.
care, and SIAT Innovation Program for Excellent Young Research- Curr. Opin. Chem. Biol. 2003, 7, 626–634.
ers (201301). 17. Mohs, A. M.; Mancini, M. C.; Singhal, S.; Provenzale, J. M.;
Supporting Information Available: Supplementary methods Leyland-Jones, B.; Wang, M. D.; Nie, S. Hand-Held Spectro-
and results. This material is available free of charge via the scopic Device for In Vivo and Intraoperative Tumour
Internet at http://pubs.acs.org. Detection: Contrast Enhancement, Detection Sensitivity,
and Tissue Penetration. Anal. Chem. 2010, 82, 9058–9065.
18. Wang, L. V.; Hu, S. Photoacoustic Tomography: In Vivo
REFERENCES AND NOTES Imaging from Organelles to Organs. Science 2012, 335,
1458–1462.
1. Celli, J. P.; Spring, B. Q.; Rizvi, I.; Evans, C. L.; Samkoe, K. S.;
Verma, S.; Pogue, B. W.; Hasan, T. Imaging and Photo- 19. Gao, L.; Fei, J.; Zhao, J.; Li, H.; Cui, Y.; Li, J. Hypocrellin-
dynamic Therapy: Mechanisms, Monitoring, and Optimiza- Loaded Gold Nanocages with High Two-Photon Efficiency
tion. Chem. Rev. 2010, 110, 2795–2838. for Photothermal/Photodynamic Cancer Therapy In Vitro.
2. Lal, S.; Clare, S. E.; Halas, N. J. Nanoshell-Enabled Photo- ACS Nano 2012, 6, 8030–8040.
thermal Cancer Therapy: Impending Clinical Impact. Acc. 20. Jang, B.; Park, J. Y.; Tung, C. H.; Kim, I. H.; Choi, Y. Gold
Chem. Res. 2008, 41, 1842–1851. Nanorod-Photosensitizer Complex for Near-Infrared Fluo-
3. Huang, P.; Lin, J.; Wang, X.; Wang, Z.; Zhang, C.; He, M.; rescence Imaging and Photodynamic/Photothermal Ther-
Wang, K.; Chen, F.; Li, Z.; Shen, G.; et al. Light-Triggered apy In Vivo. ACS Nano 2011, 5, 1086–1094.
Theranostics Based on Photosensitizer-Conjugated Car- 21. Wang, J.; Zhu, G.; You, M.; Song, E.; Shukoor, M. I.; Zhang, K.;
bon Dots for Simultaneous Enhanced-Fluorescence Altman, M. B.; Chen, Y.; Zhu, Z.; Huang, C. Z.; et al. Assembly
Imaging and Photodynamic Therapy. Adv. Mater. 2012, of Aptamer Switch Probes and Photosensitizer on Gold
24, 5104–1510. Nanorods for Targeted Photothermal and Photodynamic
4. Guo, M.; Mao, H.; Li, Y.; Zhu, A.; He, H.; Yang, H.; Wang, Y.; Cancer Therapy. ACS Nano 2012, 6, 5070–5077.
Tian, X.; Ge, C.; Peng, Q.; et al. Dual Imaging-Guided 22. Kalluru, P.; Vankayala, R.; Chiang, C. S.; Hwang, K. C.
Photothermal/Photodynamic Therapy Using Micelles. Bio- Photosensitization of Singlet Oxygen and In Vivo Photo-
materials 2014, 35, 4656–4666. dynamic Therapeutic Effects Mediated by PEGylated
5. Chen, R.; Wang, X.; Yao, X.; Zheng, X.; Wang, J.; Jiang, X. W(18)O(49) Nanowires. Angew. Chem., Int. Ed. Engl. 2013,
Near-IR-Triggered Photothermal/Photodynamic Dual- 52, 12332–12336.
Modality Therapy System via Chitosan Hybrid Nano- 23. Wang, S.; Huang, P.; Nie, L.; Xing, R.; Liu, D.; Wang, Z.; Lin, J.;
spheres. Biomaterials 2013, 34, 8314–8322. Chen, S.; Niu, G.; Lu, G.; et al. Single Continuous Wave Laser
6. Peng, C. L.; Shih, Y. H.; Lee, P. C.; Hsieh, T. M.; Luo, T. Y.; Shieh, Induced Photodynamic/Plasmonic Photothermal Therapy
M. J. Multimodal Image-Guided Photothermal Therapy Using Photosensitizer-Functionalized Gold Nanostars.
Mediated by 188Re-Labeled Micelles Containing a Cyanine- Adv. Mater. 2013, 25, 3055–3061.
Type Photosensitizer. ACS Nano 2011, 5, 5594–5607. 24. Lin, J.; Wang, S.; Huang, P.; Wang, Z.; Chen, S.; Niu, G.; Li, W.;
7. Lovell, J. F.; Jin, C. S.; Huynh, E.; Jin, H.; Kim, C.; Rubinstein, He, J.; Cui, D.; Lu, G.; et al. Photosensitizer-Loaded Gold
J. L.; Chan, W. C.; Cao, W.; Wang, L. V.; Zheng, G. Porphy- Vesicles with Strong Plasmonic Coupling Effect for Imaging-
some Nanovesicles Generated by Porphyrin Bilayers for Guided Photothermal/Photodynamic Therapy. ACS Nano
Use as Multimodal Biophotonic Contrast Agents. Nat. 2013, 7, 5320–5329.
Mater. 2011, 10, 324–332. 25. Sheng, Z.; Hu, D.; Xue, M.; He, M.; Gong, P.; Cai, L. Indocya-
8. Cheng, L.; Liu, J.; Gu, X.; Gong, H.; Shi, X.; Liu, T.; Wang, C.; nine Green Nanoparticles for Theranostic Applications.
Wang, X.; Liu, G.; Xing, H.; et al. PEGylated WS(2) Nano-Micro Lett. 2013, 5, 145–150.
Nanosheets as a Multifunctional Theranostic Agent for 26. Yu, J.; Javier, D.; Yaseen, M. A.; Nitin, N.; Richards-Kortum,
In Vivo Dual-Modal CT/Photoacoustic Imaging Guided R.; Anvari, B.; Wong, M. S. Self-Assembly Synthesis, Tu-
Photothermal Therapy. Adv. Mater. 2014, 26, 1886–1893. mour Cell Targeting, and Photothermal Capabilities of

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12321


www.acsnano.org
ARTICLE
Antibody-Coated Indocyanine Green Nanocapsules. J. Am. 43. Wu, J.; Song, C.; Jiang, C.; Shen, X.; Qiao, Q.; Hu, Y. Nucleolin
Chem. Soc. 2010, 132, 1929–1938. Targeting AS1411 Modified Protein Nanoparticle for Anti-
27. Altino glu, E. I.; Russin, T. J.; Kaiser, J. M.; Barth, B. M.; Eklund, tumour Drugs Delivery. Mol. Pharmaceutics 2013, 10,
P. C.; Kester, M.; Adair, J. H. Near-Infrared Emitting 3555–3563.
Fluorophore-Doped Calcium Phosphate Nanoparticles 44. Tian, B.; Wang, C.; Zhang, S.; Feng, L.; Liu, Z. Photothermally
for In Vivo Imaging of Human Breast Cancer. ACS Nano Enhanced Photodynamic Therapy Delivered by Nano-
2008, 2, 2075–2084. Graphene Oxide. ACS Nano 2011, 5, 7000–7009.
28. Barth, B. M.; Sharma, R.; Altino glu, E. I.; Morgan, T. T.; 45. Zhen, Z.; Tang, W.; Chuang, Y. J.; Todd, T.; Zhang, W.; Lin, X.;
Shanmugavelandy, S. S.; Kaiser, J. M.; McGovern, C.; Niu, G.; Liu, G.; Wang, L.; Pan, Z.; et al. Tumour Vasculature
Matters, G. L.; Smith, J. P.; Kester, M.; et al. Bioconjugation Targeted Photodynamic Therapy for Enhanced Delivery of
of Calcium Phosphosilicate Composite Nanoparticles for Nanoparticles. ACS Nano 2014, 8, 6004–6013.
Selective Targeting of Human Breast and Pancreatic Can- 46. Dolmans, D. E.; Kadambi, A.; Hill, J. S.; Flores, K. R.; Gerber,
cers In Vivo. ACS Nano 2010, 4, 1279–1287. J. N.; Walker, J. P.; Borel Rinkes, I. H.; Jain, R. K.; Fukumura, D.
29. Barth, B. M.; Altino glu, E.; Shanmugavelandy, S. S.; Kaiser, Targeting Tumor Vasculature and Cancer Cells in Ortho-
J. M.; Crespo-Gonzalez, D.; DiVittore, N. A.; McGovern, C.; topic Breast Tumor by Fractionated Photosensitizer Dos-
Goff, T. M.; Keasey, N. R.; Adair, J. H.; et al. Targeted ing Photodynamic Therapy. Cancer Res. 2002, 62, 4289–
Indocyanine-Green-Loaded Calcium Phosphosilicate 4294.
Nanoparticles for In Vivo Photodynamic Therapy of Leu-
kemia. ACS Nano 2011, 5, 5325–5337.
30. Zheng, C.; Zheng, M.; Gong, P.; Jia, D.; Zhang, P.; Shi, B.;
Sheng, Z.; Ma, Y.; Cai, L. Indocyanine Green-Loaded Bio-
degradable Tumour Targeting Nanoprobes for In Vitro and
In Vivo Imaging. Biomaterials 2012, 33, 5603–5609.
31. Zheng, M.; Yue, C.; Ma, Y.; Gong, P.; Zhao, P.; Zheng, C.;
Sheng, Z.; Zhang, P.; Wang, Z.; Cai, L. Single-Step Assembly
of DOX/ICG Loaded LipidPolymer Nanoparticles for
Highly Effective Chemo-Photothermal Combination Ther-
apy. ACS Nano 2013, 7, 2056–2067.
32. Zheng, M.; Zhao, P.; Luo, Z.; Gong, P.; Zheng, C.; Zhang, P.;
Yue, C.; Gao, D.; Ma, Y.; Cai, L. Robust ICG Theranostic
Nanoparticles for Folate Targeted Cancer Imaging and
Highly Effective Photothermal Therapy. ACS Appl. Mater.
Interfaces 2014, 6, 6709–6716.
33. Zhao, P.; Zheng, M.; Yue, C.; Luo, Z.; Gong, P.; Gao, G.;
Sheng, Z.; Zheng, C.; Cai, L. Improving Drug Accumulation
and Photothermal Efficacy in Tumour Depending on Size
of ICG Loaded Lipid-Polymer Nanoparticles. Biomaterials
2014, 35, 6037–6046.
34. Hannah, A.; Luke, G.; Wilson, K.; Homan, K.; Emelianov, S.
Indocyanine Green-Loaded Photoacoustic Nanodroplets:
Dual Contrast Nanoconstructs for Enhanced Photoacous-
tic and Ultrasound Imaging. ACS Nano 2014, 8, 250–259.
35. Wu, L.; Fang, S.; Shi, S.; Deng, J.; Liu, B.; Cai, L. Hybrid
Polypeptide Micelles Loading Indocyanine Green for
Tumour Imaging and Photothermal Effect Study. Bio-
macromolecules 2013, 14, 3027–3033.
36. Liu, P.; Yue, C.; Shi, B.; Gao, G.; Li, M.; Wang, B.; Ma, Y.; Cai, L.
Dextran Based Sensitive Theranostic Nanoparticles for
Near-Infrared Imaging and Photothermal Therapy In Vitro.
Chem. Commun. 2013, 49, 6143–6145.
37. Sharma, P.; Bengtsson, N. E.; Walter, G. A.; Sohn, H. B.; Zhou,
G.; Iwakuma, N.; Zeng, H.; Grobmyer, S. R.; Scott, E. W.;
Moudgil, B. M. Gadolinium-Doped Silica Nanoparticles
Encapsulating Indocyanine Green for Near Infrared and
Magnetic Resonance Imaging. Small 2012, 8, 2856–2868.
38. Moody, E. D.; Viskari, P. J.; Colyer, C. L. Non-Covalent
Labeling of Human Serum Albumin with Indocyanine
Green: a Study by Capillary Electrophoresis with Diode
Laser-Induced Fluorescence Detection. J. Chromatogr. B:
Biomed. Sci. Appl. 1999, 729, 55–64.
39. Elzoghby, A. O.; Samy, W. M.; Elgindy, N. A. Albumin-Based
Nanoparticles as Potential Controlled Release Drug Deliv-
ery Systems. J. Controlled Release 2012, 157, 168–182.
40. Elsadek, B.; Kratz, F. Impact of Albumin on Drug Delivery-
New Applications on the Horizon. J. Controlled Release
2012, 157, 4–28.
41. Xu, J.; Wang, J.; Luft, J. C.; Tian, S.; Owens, G. J.; Pandya, A. A.;
Berglund, P.; Pohlhaus, P.; Maynor, B. W.; Smith, J.; et al.
Rendering Protein-Based Particles Transiently Insoluble
for Therapeutic Applications. J. Am. Chem. Soc. 2012,
134, 8774–8777.
42. Riener, C. K.; Kada, G.; Gruber, H. J. Quick Measurement
of Protein Sulfhydryls with Ellman's Reagent and with
4,40 -dithiodipyridine. Anal. Bioanal. Chem. 2002, 373,
266–276.

SHENG ET AL. VOL. 8 ’ NO. 12 ’ 12310–12322 ’ 2014 12322


www.acsnano.org

You might also like